Skip to main content
Advertisement

Main menu

  • Home
  • Articles
    • Current Issue
    • Fast Forward
    • Latest Articles
    • Archive
  • Information
    • Instructions to Authors
    • Submit a Manuscript
    • FAQs
    • For Subscribers
    • Terms & Conditions of Use
    • Permissions
  • Editorial Board
  • Alerts
    • Alerts
    • RSS Feeds
  • Virtual Issues
  • Feedback
  • Other Publications
    • Drug Metabolism and Disposition
    • Journal of Pharmacology and Experimental Therapeutics
    • Molecular Pharmacology
    • Pharmacological Reviews
    • Pharmacology Research & Perspectives
    • ASPET

User menu

  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
Journal of Pharmacology and Experimental Therapeutics
  • Other Publications
    • Drug Metabolism and Disposition
    • Journal of Pharmacology and Experimental Therapeutics
    • Molecular Pharmacology
    • Pharmacological Reviews
    • Pharmacology Research & Perspectives
    • ASPET
  • My alerts
  • Log in
  • My Cart
Journal of Pharmacology and Experimental Therapeutics

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Fast Forward
    • Latest Articles
    • Archive
  • Information
    • Instructions to Authors
    • Submit a Manuscript
    • FAQs
    • For Subscribers
    • Terms & Conditions of Use
    • Permissions
  • Editorial Board
  • Alerts
    • Alerts
    • RSS Feeds
  • Virtual Issues
  • Feedback
  • Visit jpet on Facebook
  • Follow jpet on Twitter
  • Follow jpet on LinkedIn
Research ArticleDrug Discovery and Translational Medicine

Positive Allosteric Modulation of the Muscarinic M1 Receptor Improves Efficacy of Antipsychotics in Mouse Glutamatergic Deficit Models of Behavior

Kwok H. C. Choy, David M. Shackleford, Daniel T. Malone, Shailesh N. Mistry, Rahul T. Patil, Peter J. Scammells, Christopher J. Langmead, Christos Pantelis, Patrick M. Sexton, Johnathan R. Lane and Arthur Christopoulos
Journal of Pharmacology and Experimental Therapeutics November 2016, 359 (2) 354-365; DOI: https://doi.org/10.1124/jpet.116.235788
Kwok H. C. Choy
Drug Discovery Biology (K.H.C.C., D.T.M, C.J.L, P.M.S, J.R.L, A.C.), Centre for Drug Candidate Optimization (D.M.S., R.T.P.), and Medicinal Chemistry (S.N.M, P.J.S.), Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia; and Melbourne Neuropsychiatry Centre, Department of Psychiatry and Centre for Neural Engineering, University of Melbourne, Melbourne, Australia (C.P.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Kwok H. C. Choy
David M. Shackleford
Drug Discovery Biology (K.H.C.C., D.T.M, C.J.L, P.M.S, J.R.L, A.C.), Centre for Drug Candidate Optimization (D.M.S., R.T.P.), and Medicinal Chemistry (S.N.M, P.J.S.), Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia; and Melbourne Neuropsychiatry Centre, Department of Psychiatry and Centre for Neural Engineering, University of Melbourne, Melbourne, Australia (C.P.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Daniel T. Malone
Drug Discovery Biology (K.H.C.C., D.T.M, C.J.L, P.M.S, J.R.L, A.C.), Centre for Drug Candidate Optimization (D.M.S., R.T.P.), and Medicinal Chemistry (S.N.M, P.J.S.), Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia; and Melbourne Neuropsychiatry Centre, Department of Psychiatry and Centre for Neural Engineering, University of Melbourne, Melbourne, Australia (C.P.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Shailesh N. Mistry
Drug Discovery Biology (K.H.C.C., D.T.M, C.J.L, P.M.S, J.R.L, A.C.), Centre for Drug Candidate Optimization (D.M.S., R.T.P.), and Medicinal Chemistry (S.N.M, P.J.S.), Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia; and Melbourne Neuropsychiatry Centre, Department of Psychiatry and Centre for Neural Engineering, University of Melbourne, Melbourne, Australia (C.P.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Rahul T. Patil
Drug Discovery Biology (K.H.C.C., D.T.M, C.J.L, P.M.S, J.R.L, A.C.), Centre for Drug Candidate Optimization (D.M.S., R.T.P.), and Medicinal Chemistry (S.N.M, P.J.S.), Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia; and Melbourne Neuropsychiatry Centre, Department of Psychiatry and Centre for Neural Engineering, University of Melbourne, Melbourne, Australia (C.P.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Peter J. Scammells
Drug Discovery Biology (K.H.C.C., D.T.M, C.J.L, P.M.S, J.R.L, A.C.), Centre for Drug Candidate Optimization (D.M.S., R.T.P.), and Medicinal Chemistry (S.N.M, P.J.S.), Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia; and Melbourne Neuropsychiatry Centre, Department of Psychiatry and Centre for Neural Engineering, University of Melbourne, Melbourne, Australia (C.P.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Christopher J. Langmead
Drug Discovery Biology (K.H.C.C., D.T.M, C.J.L, P.M.S, J.R.L, A.C.), Centre for Drug Candidate Optimization (D.M.S., R.T.P.), and Medicinal Chemistry (S.N.M, P.J.S.), Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia; and Melbourne Neuropsychiatry Centre, Department of Psychiatry and Centre for Neural Engineering, University of Melbourne, Melbourne, Australia (C.P.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Christos Pantelis
Drug Discovery Biology (K.H.C.C., D.T.M, C.J.L, P.M.S, J.R.L, A.C.), Centre for Drug Candidate Optimization (D.M.S., R.T.P.), and Medicinal Chemistry (S.N.M, P.J.S.), Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia; and Melbourne Neuropsychiatry Centre, Department of Psychiatry and Centre for Neural Engineering, University of Melbourne, Melbourne, Australia (C.P.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Patrick M. Sexton
Drug Discovery Biology (K.H.C.C., D.T.M, C.J.L, P.M.S, J.R.L, A.C.), Centre for Drug Candidate Optimization (D.M.S., R.T.P.), and Medicinal Chemistry (S.N.M, P.J.S.), Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia; and Melbourne Neuropsychiatry Centre, Department of Psychiatry and Centre for Neural Engineering, University of Melbourne, Melbourne, Australia (C.P.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Johnathan R. Lane
Drug Discovery Biology (K.H.C.C., D.T.M, C.J.L, P.M.S, J.R.L, A.C.), Centre for Drug Candidate Optimization (D.M.S., R.T.P.), and Medicinal Chemistry (S.N.M, P.J.S.), Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia; and Melbourne Neuropsychiatry Centre, Department of Psychiatry and Centre for Neural Engineering, University of Melbourne, Melbourne, Australia (C.P.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Arthur Christopoulos
Drug Discovery Biology (K.H.C.C., D.T.M, C.J.L, P.M.S, J.R.L, A.C.), Centre for Drug Candidate Optimization (D.M.S., R.T.P.), and Medicinal Chemistry (S.N.M, P.J.S.), Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia; and Melbourne Neuropsychiatry Centre, Department of Psychiatry and Centre for Neural Engineering, University of Melbourne, Melbourne, Australia (C.P.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • eLetters
  • PDF + SI
  • PDF
Loading

Abstract

Current antipsychotics are effective in treating the positive symptoms associated with schizophrenia, but they remain suboptimal in targeting cognitive dysfunction. Recent studies have suggested that positive allosteric modulation of the M1 muscarinic acetylcholine receptor (mAChR) may provide a novel means of improving cognition. However, very little is known about the potential of combination therapies in extending coverage across schizophrenic symptom domains. This study investigated the effect of the M1 mAChR positive allosteric modulator BQCA [1-(4-methoxybenzyl)-4-oxo-1,4-dihydroquinoline-3-carboxylic acid], alone or in combination with haloperidol (a first-generation antipsychotic), clozapine (a second-generation atypical antipsychotic), or aripiprazole (a third-generation atypical antipsychotic), in reversing deficits in sensorimotor gating and spatial memory induced by the N-methyl-d-aspartate receptor antagonist, MK-801 [(5R,10S)-(+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine]. Sensorimotor gating and spatial memory induction are two models that represent aspects of schizophrenia modeled in rodents. In prepulse inhibition (an operational measure of sensorimotor gating), BQCA alone had minimal effects but exhibited different levels of efficacy in reversing MK-801–induced prepulse inhibition disruptions when combined with a subeffective dose of each of the three (currently prescribed) antipsychotics. Furthermore, the combined effect of BQCA and clozapine was absent in M1−/− mice. Interestingly, although BQCA alone had no effect in reversing MK-801–induced memory impairments in a Y-maze spatial test, we observed a reversal upon the combination of BQCA with atypical antipsychotics, but not with haloperidol. These findings provide proof of concept that a judicious combination of existing antipsychotics with a selective M1 mAChR positive allosteric modulator can extend antipsychotic efficacy in glutamatergic deficit models of behavior.

Introduction

Schizophrenia affects approximately 1% of the population and is characterized by positive, negative, and cognitive symptoms (Pantelis et al., 1997, 1999, 2001; Grube et al., 1998; Harvey et al., 1998; Heinrichs and Zakzanis, 1998; Kandel, 2000; Bora et al., 2009). Current antipsychotics are generally successful in treating the positive symptoms (e.g., hallucinations and delusions) but remain largely ineffective in relieving negative and cognitive symptoms and also possess side effects (Speller et al., 1997; Pantelis and Lambert, 2003; Lieberman et al., 2005; Keefe et al., 2007; Gao et al., 2008; Nasrallah, 2008; Sakurai et al., 2013). Hence, there is an unmet need for newer approaches that could target additional symptom domains and produce fewer side effects.

Existing antipsychotics treat positive schizophrenic symptoms by targeting the dopamine system, particularly the D2 receptor (Conn et al., 2008; Miyamoto et al., 2012). The broader, poorly treated, negative and cognitive domains involve additional pharmacological targets. Glutamatergic neurotransmission, in particular that mediated by the N-methyl-d-aspartate receptor (NMDAR), is potentially one such target (Rujescu et al., 2006; Javitt, 2007; Stone et al., 2007; Gordon, 2010). NMDAR antagonists, such as phencyclidine, produce psychotic-like symptoms in humans (Javitt and Zukin, 1991; Murray, 2002; Moghaddam and Javitt, 2012). Subanesthetic doses of MK-801 [(5R,10S)-(+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine], a more potent NMDAR antagonist, also induce animal behaviors that resemble aspects of schizophrenia, such as deficits of gating mechanisms and spatial memory (Bubeníková et al., 2005; Long et al., 2006; Bradford et al., 2010; van der Staay et al., 2011; Bubser et al., 2014). Accordingly, the ability of novel compounds to reduce MK-801–induced behaviors is commonly used as part of a battery of tests that assess preclinical utility of investigational molecules as potential antipsychotics (Enomoto et al., 2008; Bubser et al., 2014; Park et al., 2014).

Muscarinic acetylcholine receptors (mAChRs), in particular central M1 and M4 mAChRs, are implicated in learning and memory and have been pursued as therapeutic targets for cognitive dysfunction, including in schizophrenia (Langmead et al., 2008; Lieberman et al., 2008). Furthermore, cholinergic and dopaminergic networks can interact with one another (Lester et al., 2010). For example, stimulation of mAChRs in the ventral tegmental area increases dopamine release in the striatum and frontal cortex (Gronier et al., 2000), and knockout of M1 mAChRs increases striatal dopamine levels (Gerber et al., 2001). Importantly, the cholinergic system also interacts with glutamatergic transmission; M1 mAChRs modulate NMDAR current in hippocampal neurons (Jones et al., 2008). Of note, xanomeline, an M1 and M4 mAChR-preferring agonist, improved psychotic symptoms in patients with Alzheimer disease and cognitive function in people with schizophrenia (Bodick et al., 1997; Shekhar et al., 2008). However, further clinical development of xanomeline was precluded due to unacceptable side effects, predominantly reflecting off-target activity via the highly conserved orthosteric site at other mAChR subtypes (Bodick et al., 1997; Wood et al., 1999; Bymaster et al., 2002). Encouragingly, mAChRs possess spatially distinct allosteric sites that offer alternative strategies for greater selectivity (Christopoulos, 2002; Bridges et al., 2010; Bolbecker and Shekhar, 2012; Conn et al., 2014; Kruse et al., 2014; Thal et al., 2016). BQCA [1-(4-methoxybenzyl)-4-oxo-1,4-dihydroquinoline-3-carboxylic acid], a highly selective M1 mAChR positive allosteric modulator (PAM) with no reported off-target activities, reversed scopolamine-disrupted memory, rescued memory deficits in a mouse model of Alzheimer disease, and also had procognitive effects on baseline memory performance (Ma et al., 2009; Shirey et al., 2009; Chambon et al., 2011, 2012; Galloway et al., 2014). The effects may be due to activation of neurons in the prefrontal cortex and hippocampus, as these regions express high levels of M1 mAChRs (Porter et al., 2002; Ma et al., 2009).

The superior efficacy of existing atypical antipsychotics may be related to their complex polypharmacology (Miyamoto et al., 2005). However, less is known about the potential of combination therapies for improved treatment of negative and cognitive schizophrenic domains. Most atypical antipsychotics have limited affinity for the M1 mAChR; however, a few (e.g., clozapine and olanzapine) are antagonists at the M1 mAChR, an action linked to acute memory impairment (Bymaster et al., 1999; Didriksen et al., 2006; Mutlu et al., 2011; Miyamoto et al., 2012). Hence, coadministration of a compound that could specifically activate or potentiate the M1 mAChR together with select current antipsychotics may offer additional therapeutic value in extending antipsychotic efficacy. This study thus aimed to investigate the effects of a combination of BQCA with selected typical and atypical antipsychotics on MK-801–induced behaviors in mice, specifically prepulse inhibition (PPI) disruption and impaired memory in the Y-maze spatial test.

Materials and Methods

Materials.

Chinese hamster ovary (CHO) FlpIn cells and Dulbecco’s modified Eagle’s medium were purchased from Invitrogen (Carlsbad, CA). Fetal bovine serum was purchased from ThermoTrace (Melbourne, Australia). Hygromycin-B was purchased from Roche (Mannheim, Germany). N-[3H] methylscopolamine ([3H]NMS; specific activity, 85 Ci/mmol) and Ultima gold scintillation liquid were purchased from PerkinElmer Life Sciences (Boston, MA). BQCA, clozapine, and aripiprazole were synthesized in-house, and all other chemicals were purchased from Sigma-Aldrich (St. Louis, MO). FlpIn CHO cells stably expressing the human M1 mAChR were generated and maintained as described previously (Abdul-Ridha et al., 2014).

In Vitro Competition Binding Assays.

FlpIn CHO cells expressing the human M1 mAChR were plated at 25,000 cells per well in 96-well Isoplates (PerkinElmer Life Sciences). The next day, cells were incubated in a final volume of 100 μl HEPES buffer (10 mM HEPES, 145 mM NaCl, 1 mM MgSO4⋅7H2O, 10 mM glucose, 5 mM KCl, 2 mM CaCl2, and 1.5 mM NaHCO3, pH 7.4) containing increasing concentrations of a competing cold ligand carbachol (in the absence or presence of 10 μM BQCA) in the presence of 0.1 nM [3H]NMS for 4 hours at 4°C (to avoid potential confounding effects of competing agonist ligands on receptor internalization while ensuring reactions reached equilibrium). Nonspecific binding was defined in the presence of 100 μM atropine. For all experiments, termination of the assay was performed by rapid removal of the radioligand followed by two 100-μl washes with ice-cold 0.9% NaCl buffer. Radioactivity was determined by addition of 100 μl Microscint scintillation liquid (PerkinElmer Life Sciences) to each well and counting in a MicroBeta plate reader (PerkinElmer Life Sciences).

Animals and Drugs.

The experiments were performed using 2- to 4-month-old male C57Bl/6J, M1 mAChR homozygous knockout (M1−/−), and wild-type (M1+/+) mice in accordance with the Australian Code of Practice for the Care and Use of Animals for Scientific Purposes. Behavioral testing procedures were approved by the Animal Ethics Committee of the Monash Institute of Pharmaceutical Sciences. The M1−/− mice, which were backcrossed to C57Bl/6N for more than 11 generations, and both M1+/+ and M1−/− mice shared the same C57Bl/6N background (Bymaster et al., 2003; Thomsen et al., 2010). They were bred and genotyped at the Monash Institute of Pharmaceutical Sciences (Parkville, Australia). Mice were housed on a reversed light/dark cycle (lights on 7 PM) with ad libitum access to food and water. As outlined in the Results and in Table 1, two different routes of administration were used: C57Bl/6J mice received BQCA via subcutaneous injection, as per a previous study in this mouse strain from our laboratory when investigating another class of mAChR PAM (Suratman et al., 2011), and were tested in both PPI and Y-maze behavioral tests. However, prior studies have shown that M1+/+ and M1−/− mice on a C57Bl/6N substrain background can exhibit a different PPI profile (Matsuo et al., 2010) compared with C57Bl/6J mice. Given the limited number of animals available for these latter studies and our desire to maximize interanimal consistency, we performed new pharmacokinetic studies that revealed that intraperitoneal injection of BQCA (in a solubilized formulation) resulted in higher exposure in this instance (see Results). BQCA (1–20 mg/kg) was suspended in a combination of 50% Pharmasolve (Ashland Inc., Bridgewater, NJ) and 1.1% Tween 20 (Sigma-Aldrich, Castle Hill, Australia) in injection water (PLP, Glendenning, Australia) for subcutaneous injection (Suratman et al., 2011), or dissolved in 15% dimethylsulfoxide (DMSO) (Sigma-Aldrich), 2% Tween 20, and 22 mM Tris buffer (pH 8.9) for intraperitoneal injection. Together with BQCA administration (subcutaneously or intraperitoneally), animals received a single intraperitoneal dose of 0.03–0.25 mg/kg haloperidol (Tocris, Bristol, UK) (dissolved in 1% DMSO saline), 0.5–2 mg/kg clozapine, or 2.5 mg/kg aripiprazole (dissolved in 2% Tween 20 saline). Twenty-five minutes later, mice received an intraperitoneal injection of 0.15 or 0.3 mg/kg MK-801 (Sigma-Aldrich) dissolved in 0.1% ascorbic acid saline, and mice were then subjected to PPI or a Y-maze training session (Ytrain) 20 minutes afterward (see Results and figures for timelines). The doses of antipsychotics were specifically chosen to be subeffective in the model under test, based on previous studies (haloperidol; Bast et al., 2000), or pilot studies conducted in-house (clozapine and aripiprazole). Our pilot study showed that 4 mg/kg clozapine, or 20 mg/kg aripiprazole, reversed PPI disruption induced by 0.3 mg/kg MK-801. Haloperidol, at any doses below those that cause catalepsy (i.e., <0.5 mg/kg, above which catalepsy is observed), was ineffective at reversing the MK-801 effect. In addition, all antipsychotic doses used were either lower than (aripiprazole and clozapine) or within (haloperidol) the clinically comparable range with regard to their likely dopamine D2 receptor occupancies in humans (Kapur et al., 2003; Natesan et al., 2011).

View this table:
  • View inline
  • View popup
TABLE 1

Experimental groups and reports of ANOVA analysis

Assessment of BQCA Exposure.

BQCA exposure was assessed in three separate pharmacokinetic studies (see Results). Separate groups of C57Bl/6J, M1+/+, and M1−/− behavioral naïve mice were administered BQCA (subcutaneously or intraperitoneally), with antipsychotics (2 mg/kg clozapine, 0.25 mg/kg haloperidol, or 2.5 mg/kg aripiprazole), or vehicle and were euthanized at time points relevant to the collection of behavioral data (i.e., 45, 90, or 180 minutes postdose). Concentrations of BQCA or antipsychotics, in plasma and/or brain homogenate, were determined via ultra-performance liquid chromatography/mass spectrometry. The concentration of BQCA in the brain parenchyma (Cbrain) was calculated on the basis of the measured concentration in the brain homogenate (Cbrain homogenate), after correcting for the contribution of compound contained within the vascular space of brain samples as follows:Embedded ImageCbrain, Cbrain homogenate, and Cbrain vasculature are the concentrations of compound in the brain parenchyma, brain homogenate, and brain vasculature, respectively (in nanograms per gram). Cplasma is the concentration of compound in plasma (in nanograms per milliliter). Vp is the brain plasma volume (0.017 µl/g for C57Bl6 mice; Nicolazzo et al., 2010).

The unbound concentration of BQCA (Cunbound) in the brain parenchyma was then calculated as Ctotal × fu, using published fraction free concentration (fu) values for BQCA (i.e., fu,brain = 0.126; Gould et al., 2015). Otherwise, the total concentration of antipsychotics (Ctotal) is presented. The data are expressed in nanomolar units assuming a brain density of 1 g/ml.

PPI Test.

PPI was performed in a sound-attenuated room using SR-LAB startle chambers (San Diego Instruments, San Diego, CA). Mice subjected to PPI testing received BQCA (1–20 mg/kg) with or without antipsychotics, and then MK-801 (0.3 mg/kg) or its vehicle. Each PPI session lasted for 1 hour and consisted of three startle pulses (p100, p110, and p120) and was also preceded with 6-, 12-, and 18-db prepulses above a 65-db background for each pulse (referred to as pp6, pp12, and pp18, respectively), to provide a more comprehensive characterization of PPI (Yee et al., 2005). In the first PPI study conducted in C57Bl/6J mice, all treatment groups were pseudo-randomized on each testing day, with at least one vehicle– and MK-801–only treated mouse included as a control per PPI testing day. There were 15–17 mice for the vehicle-only or MK-801–only treatment groups, and there were 9–10 mice for all other treatment groups. The second PPI study was conducted in both M1+/+ and M1−/− mice using the same PPI protocol, with 13–16 mice for the vehicle and MK-801 controls and 8–9 mice for the other treatment groups. In these PPI studies, the drug effect on the overall PPI measured at the three startle pulses (i.e., p100, p110, and p120) was found to be similar; hence, only PPI data analysis obtained from p120 is reported. To simplify data presentation, the average PPI (%) is presented in the figures, and PPI (%) measured at each prepulse (pp6, pp12, and pp18) is detailed in Table 2. There was no change to the startle response in mice, except for those treated with the highest dose (20 mg/kg) of BQCA alone (in the presence of MK-801) (Supplemental Fig. 1). The average PPI data of p100 and p110 are presented in Supplemental Fig. 2.

View this table:
  • View inline
  • View popup
TABLE 2

PPI measured with prepulses at 6, 12, and 18 db above the background in C57Bl/6J mice, which received BQCA via the subcutaneous route

Data are presented as means ± S.E.M.

Y-Maze Test.

Mice received BQCA, clozapine, haloperidol, aripiprazole, or a combination of BQCA and an antipsychotic before MK-801 (0.15 mg/kg) injection. Since a lower dose of MK-801 (compared with 0.3 mg/kg used in PPI) was sufficient to disrupt spatial memory formation in the Y-maze, the dose range of BQCA chosen was also reduced to 5–10 mg/kg. The ranges of the antipsychotics were also reduced relative to those used in the PPI studies: haloperidol, 0.03 to 0.06 mg/kg; clozapine, 0.5 to 1 mg/kg; and aripiprazole, 0.125 to 0.25 mg/kg. Higher doses of aripiprazole (i.e., >0.25 mg/kg) were excluded, because mice had significantly lower locomotor activity in both training and testing sessions. Y-maze experiments were conducted in a manner similar to those previously described with 10–13 mice per treatment group (Dellu et al., 1992; Choy et al., 2008; Shipton et al., 2014). In brief, mice were allowed to explore two arms, with the novel arm closed, in a training session (Ytrain) that lasted for 10 minutes. Two hours later, the same mice were allowed to explore all three arms for 5 minutes in the testing session (Ytest). Behavior was recorded and analyzed with video tracking software (Viewer III; Biobserve GmbH, Bonn, Germany), and the time spent in each arm, during the testing session, was used as a measure of the exploratory preferences of mice. A significant difference between novel and familiar arms was considered as a measure of baseline memory function. In a preliminary study, prior to the full Y-maze test, MK-801 disrupted memory function when it was administered before the training session rather than after (data not shown). This finding is consistent with a previous study in which MK-801 disrupted memory in a novel objection recognition test in mice (Nilsson et al., 2007), suggesting that NMDARs blocked by MK-801 disrupt memory acquisition during the training phase. The Y-maze data of mice treated with the same vehicle used to administer MK-801 (i.e., 0.1% ascorbic acid in saline) and a combination of BQCA and antipsychotics are also reported in Supplemental Fig. 3.

Data Analysis.

For the in vitro studies, competition binding parameters were determined via nonlinear regression as described previously (Canals et al., 2012). All affinity values were estimated as logarithms, and statistical comparisons between values were performed by t tests using GraphPad Prism software (version 6.0; GraphPad Software Inc., La Jolla, CA). Behavioral data were analyzed using IBM SPSS (version 22.0; IBM, Armonk, NY). PPI (%) was analyzed with analysis of variance (ANOVA) with repeated measures of prepulses (pp6, pp12, and pp18), with drug treatments as main factors. Tukey’s post hoc test was then used to determine the level of significance at each prepulse and average values of PPI (%) among groups when there was a main drug effect or an interaction. For Y-maze data, the time spent in novel and familiar arms is presented (in seconds). If the overall main arm effect was significant in each cohort, pairwise ANOVA comparisons were performed between the novel and familiar arms in each group. Both PPI and Y-maze data analysis were divided into BQCA alone, BQCA plus haloperidol, BQCA plus clozapine, and BQCA plus aripiprazole cohorts and included their vehicle control in the ANOVA analysis. The drug effects and interactions after the ANOVA analysis are reported in Table 1, and post hoc tests are reported in the Results and/or indicated in the figures. Data are presented as means ± S.E.M. A value of P < 0.05 was considered statistically significant.

Results

BQCA, But Not Typical or Atypical Antipsychotics, Acts as a PAM of Acetylcholine at the M1 mAChR.

BQCA is a highly selective PAM of acetylcholine (ACh) binding and function at the M1 mAChR (Ma et al., 2009; Canals et al., 2012). Therefore, any effect of BQCA in vivo is most likely mediated by its interaction with the M1 mAChR. Nonetheless, to ensure that there is no specific on-target allosteric interaction between BQCA and the selected antipsychotics, we performed competition-binding experiments using the radioligand [3H]NMS on CHO FlpIn cells stably expressing the human M1 mAChR. As expected, clozapine displayed submicromolar affinity for the M1 mAChR (Ki = 0.3 μM; Fig. 1A). In contrast, neither haloperidol nor aripiprazole was able to fully inhibit [3H]NMS binding, even at a concentration of 100 μM. Although 10 μM BQCA caused a significant (60-fold) increase in ACh affinity, in agreement with our previous findings (Abdul-Ridha et al., 2014), BQCA had no effect on the affinity of clozapine (Fig. 1B); BQCA also had no effect on haloperidol and aripiprazole (data not shown). Therefore, BQCA does not modulate the binding of the selected antipsychotics at the M1 mAChR.

Fig. 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 1.

BQCA does not allosterically modulate antipsychotics at the M1 mAChR. (A) Antipsychotic affinity was measured at the human M1 mAChR stably expressed in CHO cells using competition binding experiments with the antagonist [3H]NMS. Clozapine (pKB = 6.46 ± 0.10) displayed significant affinity for the M1 mAChR. In contrast, even at a maximal concentration of 100 μM, haloperidol and aripiprazole were unable to completely displace [3H]NMS. (B) Competition binding studies between ACh or clozapine with [3H]NMS were performed in the absence (open bars) and presence (filled bars) of 10 μM BQCA. BQCA (10 μM) caused a significant 60-fold increase in ACh affinity (P < 0.001, two-tailed t test) but had no significant effect on the affinity of clozapine.

Brain Exposure of BQCA.

The apparent brain/plasma ratio from our in vivo exposure study (data not shown) was consistent with that observed previously (Ma et al., 2009; Shirey et al., 2009; Gould et al., 2015). Also, there was a clear dose-dependent increase in brain exposure in all cohorts of mice with BQCA administrated via subcutaneous or intraperitoneal injection (Supplemental Fig. 4A). Of note, all C57Bl/6J mice used in the behavioral studies reported in the main text received BQCA via the subcutaneous route. In the last PPI study using M1+/+ and M1−/− mice (see below), the animals received BQCA dissolved in DMSO-based vehicle via intraperitoneal injection, as in vivo exposure data indicated that this method of administration resulted in higher overall exposure of BQCA in this substrain (Supplemental Fig. 4B). Importantly, as shown in Supplemental Fig. 4C, in no instance was the brain exposure of either BQCA or any of the antipsychotics increased when mice were dosed with both classes of compound. Therefore, any observed behavioral effects of the combination are not a result of increased exposure at their respective sites of action.

BQCA Alone Does Not Rescue MK-801–Induced Disruptions in PPI.

The main effect of MK-801 in all cohorts of mice (Table 1) confirmed a significant PPI disruption in MK-801–treated mice (Figs. 2 and 3). However, there was an absence of a BQCA effect in the BQCA-alone cohort, suggesting that BQCA alone did not have any substantial rescuing effects on PPI disruption induced by MK-801 (Fig. 2B).

Fig. 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 2.

BQCA alone has minimal effects on MK-801–induced disruptions in PPI. (A) A timeline of drug treatment and PPI testing is presented. MK-801 (0.3 mg/kg, i.p.) was administered 25 minutes after BQCA and antipsychotics, and mice were subjected to PPI testing 20 minutes after MK-801 treatment. (B) MK-801 (0.3 mg/kg, i.p.) induced significant PPI disruption in C57Bl/6J mice. BQCA (0–20 mg/kg, s.c.) did not reverse the MK-801–disrupted PPI. ^P < 0.05 (for significant MK-801– induced disruption). Data are presented as means ± S.E.M. (vehicle– and MK-801–only controls, n = 15–17, which were reused in each cohort set, in this figure and in Fig. 3; and other treatment groups, n = 9–10).

Fig. 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 3.

Combination of subeffective doses of BQCA and haloperidol or clozapine can reverse MK-801–induced disruption in average PPI. (A and B) Both haloperidol (0.25 mg/kg) and clozapine (2 mg/kg) did not reverse MK-801–induced PPI disruption; however, the combination with BQCA (20 mg/kg, s.c.) resulted in significant reversal of PPI disruption in C57Bl/6J mice. (C) The combination of BQCA (20 mg/kg, s.c.) and aripiprazole did not produce a reversal in average PPI, except PPI measured at pp18 (Table 2). *P < 0.05 (for difference with MK-801 post hoc comparisons); **P < 0.001 (for difference with MK-801 post hoc comparisons); ^P < 0.05 (for significant MK-801–induced disruption). Data are presented as means ± S.E.M. (vehicle– and MK-801–only controls, n = 15–17, which were reused in each cohort set, in this figure and in Fig. 2; and other treatment groups, n = 9–10).

Combination of Subeffective Doses of BQCA with Typical or Atypical Antipsychotics Produces Significant Reversals in MK-801–Induced Disruptions in PPI.

As indicated in the Materials and Methods, the dose of haloperidol (0.25 mg/kg) was specifically chosen from prior studies to represent a subeffective dose level in the PPI studies. In the BQCA plus haloperidol cohort, there was a significant BQCA × haloperidol effect (Table 1) as an indication of a synergistic effect between the two compounds. Post hoc analysis (Table 2) showed that MK-801–induced disruption in PPI was not fully reversed by either haloperidol alone or a combination of haloperidol with 1–10 mg/kg BQCA. However, a complete reversal of MK-801–induced PPI disruption was observed when the subeffective dose of haloperidol was combined with 20 mg/kg BQCA (Fig. 3A). Similarly, clozapine (2 mg/kg) alone and in combination with 1–10 mg/kg BQCA failed to fully reverse MK-801–induced PPI disruption. However, a significant reversal was observed with the combination of clozapine and BQCA (20 mg/kg) (Fig. 3B), although there was an absence of BQCA by clozapine interaction in the ANOVA, indicating that both BQCA and clozapine were likely behaving additively rather than synergistically. Comparable trends were also observed in the aripiprazole plus BQCA cohort, but only at the highest prepulse tested (i.e., pp18) (Fig. 3C; Table 2). In this instance, the significant prepulse by BQCA by aripiprazole interaction (Table 1) indicated the presence of a synergistic effect of PPI measured at pp18. Taken together, these results reveal that BQCA enhanced subeffective doses of commonly used antipsychotics to produce a rescue of PPI deficits in MK-801–treated mice.

BQCA Enhanced the Rescuing Effect of Clozapine or Aripiprazole, But Not That of Haloperidol, in the Y-Maze Test.

To explore memory performance, the duration spent (in seconds) in novel and familiar arms for each treatment group was analyzed (Table 1). There was a main arm effect and a significant arm × MK-801 interaction, indicating disruption of short-term memory in the Y-maze test by MK-801 (Table 1). Further analysis revealed differences between the novel and familiar arms in vehicle-treated mice (P = 0.002; Fig. 4B). In MK-801–treated mice, pretreatment with 10 mg/kg BQCA and 1 mg/kg clozapine (P = 0.007; Fig. 5B), or 5 mg/kg BQCA and 0.125 mg/kg aripiprazole (P = 0.026; Fig. 5C), also showed novel versus familiar arm differences, which is indicative of normal memory function as seen in vehicle control mice (Fig. 5). There were no differences in the other treatment groups, including BQCA alone or in combination with haloperidol (Fig. 5A). These results demonstrate a synergistic effect between BQCA and the atypical antipsychotics, clozapine or aripiprazole, on memory function, which was not seen with BQCA on its own or in combination with haloperidol, in this acute memory disruption model.

Fig. 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 4.

MK-801 induces spatial memory deficits in C57Bl/6J mice in the Y-maze test. (A) A timeline of drug treatment and Y-maze testing is presented. BQCA and antipsychotics were given 25 minutes prior to MK-801 (0.15 mg/kg) administration, and the mice were then subjected to a Y-maze training session 20 minutes after MK-801 treatment. The retention period was 2 hours (between training [Ytrain] and testing [Ytest] sessions). The duration spent (in seconds) in the novel and familiar arms was used as the measure of memory function. (B) Analysis showed no rescue of memory deficits induced by MK-801 (0.15 mg/kg) for mice pretreated with BQCA alone. *P < 0.05 (for difference between the novel and familiar arms as an indication of normal memory function). Data are presented as means ± S.E.M. (n = 10–13 mice per group). N, novel.

Fig. 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 5.

The combination of BQCA with clozapine or aripiprazole, but not with haloperidol, improves MK-801–induced spatial memory in C57Bl/6J mice in the Y-maze test. (A) Mice pretreated with BQCA combined with haloperidol showed no rescue of MK-801–induced memory disruption. (B and C) In contrast, mice treated with combinations of BQCA (10 mg/kg, s.c.) and clozapine (1 mg/kg), or BQCA (5 mg/kg, s.c.) and aripiprazole (0.125 mg/kg), showed rescue of the MK-801 (0.15 mg/kg) memory disruptive effect. *P < 0.05 (for difference between the novel and familiar arms as an indication of normal memory function). Data are presented as means ± S.E.M. (n = 10–13 mice per group). N, novel.

M1 mAChR Is Required for the Combined Effect of BQCA and Clozapine in Reversing PPI Deficits.

To confirm the requirement of the M1 mAChR for the observed combined effect, we repeated selected key BQCA and clozapine interaction studies in mice lacking the M1 mAChR; this also necessitated testing in different background mouse strains. The combined effect between BQCA and clozapine seen in reversing MK-801–induced PPI deficits was reproducible in C57Bl/6J mice using this administration method (Supplemental Fig. 5). Moreover, consistent with previous findings (Matsuo et al., 2010), we noted that both M1+/+ and M1−/− mice on a C57Bl/6N background exhibited a higher PPI compared with the C57Bl/6J mice used in the preceding studies. PPI disruption was induced to a similar extent by 0.3 mg/kg MK-801 in both M1+/+ and M1−/− mice. A partial, but statistically nonsignificant, reversal of the PPI deficit was observed after dosing clozapine alone, consistent with the observation in C57Bl/6J mice (Fig. 6). A qualitatively similar partial reversal was observed after M1+/+ mice received BQCA alone at 20 mg/kg, a trend that was not observed in C57Bl6J mice. This reversal failed to achieve statistical significance and was not observed in subsequent studies with further cohorts of M1+/+ mice. The most important finding, however, was that MK-801–disrupted PPI was rescued by the combination of BQCA (20 mg/kg) and clozapine (3 mg/kg) in the M1+/+ mice, and there was a complete absence of such rescue in the M1−/− mice receiving the same treatment (Fig. 6; Table 3). These results demonstrate the selectivity of BQCA in vivo and also confirm that the M1 mAChR is required in order to observe efficacy enhancement of clozapine actions.

Fig. 6.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 6.

The enhancement of efficacy of clozapine by BQCA in reversing PPI deficits is absent in M1 mAChR knockout mice. MK-801 (0.3 mg/kg) induced PPI disruption in both M1+/+ mice (top) and M1−/− mice (bottom). Despite differences in the PPI profile between C57Bl/6J and M1+/+ (C57Bl/6N background) mice, an additive effect of BQCA (20 mg/kg, i.p.) and clozapine (3 mg/kg) was still observed in the M1+/+ mice but not in the M1−/− mice. The synergetic effect was particularly significant at pp6 (Table 3). *P < 0.05 (for difference with MK-801 post hoc comparisons); ^P < 0.05 (for significant MK-801–induced disruption). Data are presented as means ± S.E.M. (M1+/+ vehicle and MK-801 controls, n = 15–16; M1+/+ treatment groups, n = 8–9; M1−/− vehicle and MK-801 controls, n = 13–15; and M1−/− treatment groups, n = 8).

View this table:
  • View inline
  • View popup
TABLE 3

PPI measured with prepulses at 6, 12, and 18 db above the background in M1+/+ and M1−/− (C57Bl/6 Ntac background) mice, which received BQCA via the intraperitoneal route

Data are presented as means ± S.E.M.

Discussion

This study provides proof of concept that the combination of an M1 mAChR PAM with different antipsychotic drugs can yield synergistic (or additive) efficacy in two animal behavioral models that are considered to reflect aspects of schizophrenia. These effects are not mediated by on-target allosteric interactions at the M1 mAChR between BQCA and the antipsychotics, but activation of the receptor is necessary because the interaction between BQCA and clozapine in reversing MK-801–induced PPI disruption is lost in M1−/− mice. If these findings can be generalized to other aspects or models of schizophrenia, then this may provide impetus for future consideration of the use of M1 mAChR PAMs as potential add-on therapies to selected existing drug regimens.

Haloperidol is a first-generation antipsychotic agent, representative of the requirement for potent dopamine D2 receptor antagonism in treating positive schizophrenic symptoms (Mansbach et al., 1988; Conn et al., 2008; Miyamoto et al., 2012). Clozapine was chosen as an atypical second-generation antipsychotic with a broad polypharmacology profile (Miyamoto et al., 2005; Nasrallah, 2008), whereas aripiprazole was selected as a third-generation agent that acts on D2 receptors as a partial agonist with a lower propensity toward adverse metabolic side effects (Nasrallah, 2008; Cui et al., 2010). None of these drugs, however, are effective at treating cognitive deficits associated with schizophrenia (Lieberman et al., 2005; Lublin et al., 2005). However, therapeutic targeting has not been limited to D2 receptors (Conn et al., 2008; Owen et al., 2016). For example, agents targeting mAChRs that are highly expressed in affected regions, such as the prefrontal cortex and hippocampus, have received attention for their potential in rescuing cognitive dysfunction (Bymaster et al., 2002; Shekhar et al., 2008; Bridges et al., 2010; Barak and Weiner, 2011; Bolbecker and Shekhar, 2012). In this study, we specifically focused on NMDAR antagonism by MK-801 as a means of modeling sensorimotor deficits and short-term memory disruption due to glutamatergic hypofunction (Gilmour et al., 2012), as opposed to pharmacological interventions such as amphetamine that model positive symptom domains but are more limited for studying negative or cognitive domains (Young et al., 2010; Katsnelson, 2014). For behavioral models, the PPI test measures sensorimotor gating in the brain, as it is impaired in patients with schizophrenia, and has been used in animals to identify and understand antipsychotic efficacies of clinically available drugs (Keith et al., 1991; Bakshi et al., 1994; Swerdlow et al., 1994; Swerdlow and Geyer, 1998; Cadenhead et al., 1999; Young et al., 2010; Moore et al., 2013). The Y-maze test is a short-term spatial memory paradigm for assessing hippocampal-dependent memory function (Conrad et al., 1996, 1997; Choy et al., 2008; Shipton et al., 2014), which is highly vulnerable to pathologic factors such as stress (Heckers et al., 1998; Harrison, 2004; Sweatt, 2004; van Erp et al., 2004, 2016; Tanskanen et al., 2005; Vakalopoulos, 2006; Velakoulis et al., 2006; Choy et al., 2008) and is impaired from the earliest stages of schizophrenia (Wood et al., 2002, 2003). Therefore, the Y-maze test has potential to provide preclinical insights into the effects of combining BQCA and antipsychotics on brain regions affected along the pathologic trajectory of schizophrenia (Heckers et al., 1998; Weinberger, 1999; Lipska and Weinberger, 2002; Harrison, 2004; Sweatt, 2004; van Erp et al., 2004; Harrison and Weinberger, 2005; Tanskanen et al., 2005; Velakoulis et al., 2006)

In general, BQCA alone had minimal effects in both behavioral models. With the exception of the highest dose of clozapine (5 mg/kg), which also affected the animal startle response, the doses of haloperidol, clozapine, and aripiprazole used in this study were insufficient to restore normal PPI after MK-801 treatment, consistent with findings from previous studies (Bakshi et al., 1994; Bast et al., 2000; Ishii et al., 2010). However, it should be noted that this subeffective dosing was necessary to reveal the effect of combination with BQCA. Lowering doses of antipsychotics also brings potential benefits, because the side-effect burden would be lessened if therapeutically effective lower doses were to be administered clinically. The fact that coadministration of the M1 mAChR PAM with each of the three classes of antipsychotic rescued the disrupted PPI response indicates that sensorimotor gating deficits can be particularly amenable to combination therapy, even in the case of clozapine, which itself has affinity for the M1 mAChR. Presumably, potentiation of endogenous ACh affinity at this receptor by BQCA, however, would reduce M1 mAChR occupancy by clozapine (and its metabolites) in preference for the full agonist, ACh. Importantly, we confirmed that the combined effect of BQCA and clozapine was reproducible in the M1+/+ mice but lost in the M1−/− mice, despite differences in the overall PPI profile between the C57Bl/6 substrains (Matsuo et al., 2010).

In addition to sensorimotor gating deficits, spatial memory dysfunction, which is mostly associated with decline of hippocampal function, has been commonly observed in patients with schizophrenia, even at early onset stages (Saykin et al., 1994; Weinberger and Gallhofer, 1997; Heckers et al., 1998; Wood et al., 2002; Bertolino et al., 2003; Egeland et al., 2003; Brewer et al., 2005; Bartholomeusz et al., 2011). In contrast with the PPI studies, the atypical antipsychotics, clozapine and aripiprazole, but not haloperidol, showed an enhancement of efficacy by BQCA in reversing MK-801–induced spatial memory disruption in the Y-maze. The hippocampus has a high expression of M1 mAChRs, which are important for memory formation in rodents (Talts et al., 1998; Anagnostaras et al., 2003; Lee and Kesner, 2003; Newell et al., 2007; Mauck et al., 2010; Peng et al., 2012). Indeed, BQCA enhances baseline spatial and object recognition memory in rodents (Chambon et al., 2011, 2012), reverses scopolamine-disrupted fear and working memory, and restores memory function in Tg2576 mice (Ma et al., 2009; Shirey et al., 2009; Chambon et al., 2012). To our knowledge, however, ours is the first study to extend these effects to rescue of MK-801–induced memory impairments, albeit in the presence of a coadministered antipsychotic. Interestingly, another recent study showed that a PAM of the M4 mAChR subtype could rescue MK-801–induced impairments in a touchscreen-based memory retrieval task (Bubser et al., 2014), and earlier work established that the cholinesterase inhibitor, donepezil, can also rescue MK-801–disrupted fear memory (Csernansky et al., 2005).

In terms of the brain regions mediating the interactions between the M1 mAChR PAM and antipsychotics in our models, it is likely that cortical and striatal regions are involved in the PPI effects, since they express the M1 mAChR (Han et al., 2008; Ma et al., 2009; Peng et al., 2012) and have been implicated in regulation of sensorimotor gating mechanisms (Swerdlow et al., 1990, 2001; Geyer et al., 2001; Braff, 2010). Although the M1 mAChR is also found in the hippocampus, it is unlikely to be as important in the observed effects, as MK-801–induced PPI disruption does not involve this region and, if anything, cholinergic activation in the hippocampus can itself lead to some PPI disruption (Caine et al., 1991, 1992; Zhang et al., 2000). In contrast, the Y-maze test is substantially hippocampal dependent (Conrad et al., 1996, 2007; Choy et al., 2010; Shipton et al., 2014). Nonetheless, the possible involvement of cortical regions in both behaviors should not be overlooked, as cortical, striatal, and hippocampal networks play an important role in higher cognitive function (Turnock and Becker, 2008; Pennartz et al., 2011; Britt et al., 2012).

An area of ongoing discussion is the issue of whether “single-target bullets” or “multifunction drugs” will lead to more effective medications for schizophrenia (Pantelis and Barnes, 1996; Roth et al., 2004; Miyamoto et al., 2012). What remains indisputable, however, is that schizophrenia is a disorder that requires drugs capable of targeting multiple symptom domains, in particular cognitive deficits. Moreover, the incidence of adverse effects remains a major problem for patients undergoing chronic treatment with existing antipsychotics (Nasrallah, 2008). Both of these issues may conceivably be addressed by judicious combination therapies. For instance, although clozapine was used as proof of concept in our study, it may not be amenable for combination therapies in practice due to its known on-target mAChR effects, whereas other atypical agents (e.g., aripiprazole or risperidone, which have minimal mAChR activity; Roth et al., 2004) may represent more appropriate candidates. Furthermore, the addition of a highly selective mAChR PAM (i.e., single-target bullet) may yield an increase in symptom domain coverage together with an existing antipsychotic (i.e., multifunction drug). Although these issues remain speculative at this time, our study shows that we can observe synergistic or additive efficacy in our models while essentially retaining normal exploratory behaviors in the Y-maze test and normal body reflex upon acoustic startle stimulation in the PPI test. Thus, in addition to offering novel approaches to targeting G protein–coupled receptors in a highly selective manner to address unmet medical needs, allosteric modulators may potentially allow extension of the clinical efficacy of existing therapeutics and warrant further study in this regard.

Acknowledgments

The authors thank Dr. Jürgen Wess for providing M1+/+ and M1−/− mice, Leigh Howard for maintaining the mouse colonies in-house, Dr. Susan Charman for helpful discussions regarding mouse exposure studies, and Drs. Jeremy Shonberg and Ben Capuano for synthesis of clozapine and aripiprazole.

Authorship Contributions

Participated in research design: Choy, Shackleford, Malone, Lane, Christopoulos.

Conducted experiments: Choy, Shackleford, Mistry, Patil, Scammells, Lane.

Performed data analysis: Choy, Shackleford, Malone, Lane.

Wrote or contributed to the writing of the manuscript: Choy, Shackleford, Malone, Mistry, Scammells, Langmead, Pantelis, Sexton, Lane, Christopoulos.

Footnotes

    • Received June 7, 2016.
    • Accepted September 13, 2016.
  • ↵1 Current affiliation: School of Pharmacy, Centre for Biomolecular Sciences, University of Nottingham, University Park, Nottingham, United Kingdom.

  • This research was supported by the National Health and Medical Research Council (NHMRC) of Australia [Project Grant APP1052304 and Program Grant APP1055134]. A.C. and C.P. are NHMRC Senior Principal Research Fellows; P.M.S. is an NHMRC Principal Research Fellow; and J.R.L. is an NHMRC R.D. Wright Biomedical Career Development Fellow and Monash University Larkins Fellow.

  • dx.doi.org/10.1124/jpet.116.235788.

  • ↵Embedded ImageThis article has supplemental material available at jpet.aspetjournals.org.

Abbreviations

ACh
acetylcholine
ANOVA
analysis of variance
BQCA
1-(4-methoxybenzyl)-4-oxo-1,4-dihydroquinoline-3-carboxylic acid
CHO
Chinese hamster ovary
DMSO
dimethylsulfoxide
mAChR
muscarinic acetylcholine receptor
MK-801
(5R,10S)-(+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine
NMDAR
N-methyl-d-aspartate receptor
[3H]NMS
N-[3H] methylscopolamine
PAM
positive allosteric modulator
PPI
prepulse inhibition
  • Copyright © 2016 by The American Society for Pharmacology and Experimental Therapeutics

References

  1. ↵
    1. Abdul-Ridha A,
    2. Lane JR,
    3. Mistry SN,
    4. López L,
    5. Sexton PM,
    6. Scammells PJ,
    7. Christopoulos A, and
    8. Canals M
    (2014) Mechanistic insights into allosteric structure-function relationships at the M1 muscarinic acetylcholine receptor. J Biol Chem 289:33701–33711.
    OpenUrlAbstract/FREE Full Text
  2. ↵
    1. Anagnostaras SG,
    2. Murphy GG,
    3. Hamilton SE,
    4. Mitchell SL,
    5. Rahnama NP,
    6. Nathanson NM, and
    7. Silva AJ
    (2003) Selective cognitive dysfunction in acetylcholine M1 muscarinic receptor mutant mice. Nat Neurosci 6:51–58.
    OpenUrlCrossRefPubMed
  3. ↵
    1. Bakshi VP,
    2. Swerdlow NR, and
    3. Geyer MA
    (1994) Clozapine antagonizes phencyclidine-induced deficits in sensorimotor gating of the startle response. J Pharmacol Exp Ther 271:787–794.
    OpenUrlAbstract/FREE Full Text
  4. ↵
    1. Barak S and
    2. Weiner I
    (2011) The M₁/M₄ preferring agonist xanomeline reverses amphetamine-, MK801- and scopolamine-induced abnormalities of latent inhibition: putative efficacy against positive, negative and cognitive symptoms in schizophrenia. Int J Neuropsychopharmacol 14:1233–1246.
    OpenUrlAbstract/FREE Full Text
  5. ↵
    1. Bartholomeusz CF,
    2. Proffitt TM,
    3. Savage G,
    4. Simpson L,
    5. Markulev C,
    6. Kerr M,
    7. McConchie M,
    8. McGorry PD,
    9. Pantelis C,
    10. Berger GE,
    11. et al.
    (2011) Relational memory in first episode psychosis: implications for progressive hippocampal dysfunction after illness onset. Aust N Z J Psychiatry 45:206–213.
    OpenUrlAbstract/FREE Full Text
  6. ↵
    1. Bast T,
    2. Zhang W,
    3. Feldon J, and
    4. White IM
    (2000) Effects of MK801 and neuroleptics on prepulse inhibition: re-examination in two strains of rats. Pharmacol Biochem Behav 67:647–658.
    OpenUrlCrossRefPubMed
  7. ↵
    1. Bertolino A,
    2. Sciota D,
    3. Brudaglio F,
    4. Altamura M,
    5. Blasi G,
    6. Bellomo A,
    7. Antonucci N,
    8. Callicott JH,
    9. Goldberg TE,
    10. Scarabino T,
    11. et al.
    (2003) Working memory deficits and levels of N-acetylaspartate in patients with schizophreniform disorder. Am J Psychiatry 160:483–489.
    OpenUrlCrossRefPubMed
  8. ↵
    1. Bodick NC,
    2. Offen WW,
    3. Levey AI,
    4. Cutler NR,
    5. Gauthier SG,
    6. Satlin A,
    7. Shannon HE,
    8. Tollefson GD,
    9. Rasmussen K,
    10. Bymaster FP,
    11. et al.
    (1997) Effects of xanomeline, a selective muscarinic receptor agonist, on cognitive function and behavioral symptoms in Alzheimer disease. Arch Neurol 54:465–473.
    OpenUrlCrossRefPubMed
  9. ↵
    1. Bolbecker AR and
    2. Shekhar A
    (2012) Muscarinic agonists and antagonists in schizophrenia: recent therapeutic advances and future directions. Handb Exp Pharmacol 208:167–190.
    OpenUrlCrossRef
  10. ↵
    1. Bora E,
    2. Yucel M, and
    3. Pantelis C
    (2009) Cognitive functioning in schizophrenia, schizoaffective disorder and affective psychoses: meta-analytic study. Br J Psychiatry 195:475–482.
    OpenUrlAbstract/FREE Full Text
  11. ↵
    1. Bradford AM,
    2. Savage KM,
    3. Jones DN, and
    4. Kalinichev M
    (2010) Validation and pharmacological characterisation of MK-801-induced locomotor hyperactivity in BALB/C mice as an assay for detection of novel antipsychotics. Psychopharmacology (Berl) 212:155–170.
    OpenUrlCrossRefPubMed
  12. ↵
    1. Braff DL
    (2010) Prepulse inhibition of the startle reflex: a window on the brain in schizophrenia. Curr Top Behav Neurosci 4:349–371.
    OpenUrlCrossRefPubMed
  13. ↵
    1. Brewer WJ,
    2. Francey SM,
    3. Wood SJ,
    4. Jackson HJ,
    5. Pantelis C,
    6. Phillips LJ,
    7. Yung AR,
    8. Anderson VA, and
    9. McGorry PD
    (2005) Memory impairments identified in people at ultra-high risk for psychosis who later develop first-episode psychosis. Am J Psychiatry 162:71–78.
    OpenUrlCrossRefPubMed
  14. ↵
    1. Bridges TM,
    2. LeBois EP,
    3. Hopkins CR,
    4. Wood MR,
    5. Jones CK,
    6. Conn PJ, and
    7. Lindsley CW
    (2010) The antipsychotic potential of muscarinic allosteric modulation. Drug News Perspect 23:229–240.
    OpenUrlCrossRefPubMed
  15. ↵
    1. Britt JP,
    2. Benaliouad F,
    3. McDevitt RA,
    4. Stuber GD,
    5. Wise RA, and
    6. Bonci A
    (2012) Synaptic and behavioral profile of multiple glutamatergic inputs to the nucleus accumbens. Neuron 76:790–803.
    OpenUrlCrossRefPubMed
  16. ↵
    1. Bubeníková V,
    2. Votava M,
    3. Horácek J,
    4. Pálenícek T, and
    5. Dockery C
    (2005) The effect of zotepine, risperidone, clozapine and olanzapine on MK-801-disrupted sensorimotor gating. Pharmacol Biochem Behav 80:591–596.
    OpenUrlCrossRefPubMed
  17. ↵
    1. Bubser M,
    2. Bridges TM,
    3. Dencker D,
    4. Gould RW,
    5. Grannan M,
    6. Noetzel MJ,
    7. Lamsal A,
    8. Niswender CM,
    9. Daniels JS,
    10. Poslusney MS,
    11. et al.
    (2014) Selective activation of M4 muscarinic acetylcholine receptors reverses MK-801-induced behavioral impairments and enhances associative learning in rodents. ACS Chem Neurosci 5:920–942.
    OpenUrlCrossRefPubMed
  18. ↵
    1. Bymaster FP,
    2. Felder C,
    3. Ahmed S, and
    4. McKinzie D
    (2002) Muscarinic receptors as a target for drugs treating schizophrenia. Curr Drug Targets CNS Neurol Disord 1:163–181.
    OpenUrlCrossRefPubMed
  19. ↵
    1. Bymaster FP,
    2. McKinzie DL,
    3. Felder CC, and
    4. Wess J
    (2003) Use of M1-M5 muscarinic receptor knockout mice as novel tools to delineate the physiological roles of the muscarinic cholinergic system. Neurochem Res 28:437–442.
    OpenUrlCrossRefPubMed
  20. ↵
    1. Bymaster FP,
    2. Nelson DL,
    3. DeLapp NW,
    4. Falcone JF,
    5. Eckols K,
    6. Truex LL,
    7. Foreman MM,
    8. Lucaites VL, and
    9. Calligaro DO
    (1999) Antagonism by olanzapine of dopamine D1, serotonin2, muscarinic, histamine H1 and alpha 1-adrenergic receptors in vitro. Schizophr Res 37:107–122.
    OpenUrlCrossRefPubMed
  21. ↵
    1. Cadenhead KS,
    2. Carasso BS,
    3. Swerdlow NR,
    4. Geyer MA, and
    5. Braff DL
    (1999) Prepulse inhibition and habituation of the startle response are stable neurobiological measures in a normal male population. Biol Psychiatry 45:360–364.
    OpenUrlCrossRefPubMed
  22. ↵
    1. Caine SB,
    2. Geyer MA, and
    3. Swerdlow NR
    (1991) Carbachol infusion into the dentate gyrus disrupts sensorimotor gating of startle in the rat. Psychopharmacology (Berl) 105:347–354.
    OpenUrlCrossRefPubMed
  23. ↵
    1. Caine SB,
    2. Geyer MA, and
    3. Swerdlow NR
    (1992) Hippocampal modulation of acoustic startle and prepulse inhibition in the rat. Pharmacol Biochem Behav 43:1201–1208.
    OpenUrlCrossRefPubMed
  24. ↵
    1. Canals M,
    2. Lane JR,
    3. Wen A,
    4. Scammells PJ,
    5. Sexton PM, and
    6. Christopoulos A
    (2012) A Monod-Wyman-Changeux mechanism can explain G protein-coupled receptor (GPCR) allosteric modulation. J Biol Chem 287:650–659.
    OpenUrlAbstract/FREE Full Text
  25. ↵
    1. Chambon C,
    2. Jatzke C,
    3. Wegener N,
    4. Gravius A, and
    5. Danysz W
    (2012) Using cholinergic M1 receptor positive allosteric modulators to improve memory via enhancement of brain cholinergic communication. Eur J Pharmacol 697:73–80.
    OpenUrlCrossRefPubMed
  26. ↵
    1. Chambon C,
    2. Wegener N,
    3. Gravius A, and
    4. Danysz W
    (2011) A new automated method to assess the rat recognition memory: validation of the method. Behav Brain Res 222:151–157.
    OpenUrlCrossRefPubMed
  27. ↵
    1. Choy KH,
    2. de Visser Y,
    3. Nichols NR, and
    4. van den Buuse M
    (2008) Combined neonatal stress and young-adult glucocorticoid stimulation in rats reduce BDNF expression in hippocampus: effects on learning and memory. Hippocampus 18:655–667.
    OpenUrlCrossRefPubMed
  28. ↵
    1. Choy KH,
    2. Dean O,
    3. Berk M,
    4. Bush AI, and
    5. van den Buuse M
    (2010) Effects of N-acetyl-cysteine treatment on glutathione depletion and a short-term spatial memory deficit in 2-cyclohexene-1-one-treated rats. Eur J Pharmacol 649:224–228.
    OpenUrlCrossRefPubMed
  29. ↵
    1. Christopoulos A
    (2002) Allosteric binding sites on cell-surface receptors: novel targets for drug discovery. Nat Rev Drug Discov 1:198–210.
    OpenUrlCrossRefPubMed
  30. ↵
    1. Conn PJ,
    2. Lindsley CW,
    3. Meiler J, and
    4. Niswender CM
    (2014) Opportunities and challenges in the discovery of allosteric modulators of GPCRs for treating CNS disorders. Nat Rev Drug Discov 13:692–708.
    OpenUrlCrossRefPubMed
  31. ↵
    1. Conn PJ,
    2. Tamminga C,
    3. Schoepp DD, and
    4. Lindsley C
    (2008) Schizophrenia: moving beyond monoamine antagonists. Mol Interv 8:99–107.
    OpenUrlCrossRefPubMed
  32. ↵
    1. Conrad CD,
    2. Galea LA,
    3. Kuroda Y, and
    4. McEwen BS
    (1996) Chronic stress impairs rat spatial memory on the Y maze, and this effect is blocked by tianeptine pretreatment. Behav Neurosci 110:1321–1334.
    OpenUrlCrossRefPubMed
  33. ↵
    1. Conrad CD,
    2. Lupien SJ,
    3. Thanasoulis LC, and
    4. McEwen BS
    (1997) The effects of type I and type II corticosteroid receptor agonists on exploratory behavior and spatial memory in the Y-maze. Brain Res 759:76–83.
    OpenUrlCrossRefPubMed
  34. ↵
    1. Conrad CD,
    2. McLaughlin KJ,
    3. Harman JS,
    4. Foltz C,
    5. Wieczorek L,
    6. Lightner E, and
    7. Wright RL
    (2007) Chronic glucocorticoids increase hippocampal vulnerability to neurotoxicity under conditions that produce CA3 dendritic retraction but fail to impair spatial recognition memory. J Neurosci 27:8278–8285.
    OpenUrlAbstract/FREE Full Text
  35. ↵
    1. Csernansky JG,
    2. Martin M,
    3. Shah R,
    4. Bertchume A,
    5. Colvin J, and
    6. Dong H
    (2005) Cholinesterase inhibitors ameliorate behavioral deficits induced by MK-801 in mice. Neuropsychopharmacology 30:2135–2143.
    OpenUrlCrossRefPubMed
  36. ↵
    1. Cui YH,
    2. Zheng Y,
    3. Yang YP,
    4. Liu J, and
    5. Li J
    (2010) Effectiveness and tolerability of aripiprazole in children and adolescents with Tourette’s disorder: a pilot study in China. J Child Adolesc Psychopharmacol 20:291–298.
    OpenUrlCrossRefPubMed
  37. ↵
    1. Dellu F,
    2. Mayo W,
    3. Cherkaoui J,
    4. Le Moal M, and
    5. Simon H
    (1992) A two-trial memory task with automated recording: study in young and aged rats. Brain Res 588:132–139.
    OpenUrlCrossRefPubMed
  38. ↵
    1. Didriksen M,
    2. Kreilgaard M, and
    3. Arnt J
    (2006) Sertindole, in contrast to clozapine and olanzapine, does not disrupt water maze performance after acute or chronic treatment. Eur J Pharmacol 542:108–115.
    OpenUrlCrossRefPubMed
  39. ↵
    1. Egeland J,
    2. Sundet K,
    3. Rund BR,
    4. Asbjørnsen A,
    5. Hugdahl K,
    6. Landrø NI,
    7. Lund A,
    8. Roness A, and
    9. Stordal KI
    (2003) Sensitivity and specificity of memory dysfunction in schizophrenia: a comparison with major depression. J Clin Exp Neuropsychol 25:79–93.
    OpenUrlCrossRefPubMed
  40. ↵
    1. Enomoto T,
    2. Ishibashi T,
    3. Tokuda K,
    4. Ishiyama T,
    5. Toma S, and
    6. Ito A
    (2008) Lurasidone reverses MK-801-induced impairment of learning and memory in the Morris water maze and radial-arm maze tests in rats. Behav Brain Res 186:197–207.
    OpenUrlCrossRefPubMed
  41. ↵
    1. Galloway CR,
    2. Lebois EP,
    3. Shagarabi SL,
    4. Hernandez NA, and
    5. Manns JR
    (2014) Effects of selective activation of M1 and M4 muscarinic receptors on object recognition memory performance in rats. Pharmacology 93:57–64.
    OpenUrlCrossRef
  42. ↵
    1. Gao K,
    2. Kemp DE,
    3. Ganocy SJ,
    4. Gajwani P,
    5. Xia G, and
    6. Calabrese JR
    (2008) Antipsychotic-induced extrapyramidal side effects in bipolar disorder and schizophrenia: a systematic review. J Clin Psychopharmacol 28:203–209.
    OpenUrlCrossRefPubMed
  43. ↵
    1. Gerber DJ,
    2. Sotnikova TD,
    3. Gainetdinov RR,
    4. Huang SY,
    5. Caron MG, and
    6. Tonegawa S
    (2001) Hyperactivity, elevated dopaminergic transmission, and response to amphetamine in M1 muscarinic acetylcholine receptor-deficient mice. Proc Natl Acad Sci USA 98:15312–15317.
    OpenUrlAbstract/FREE Full Text
  44. ↵
    1. Geyer MA,
    2. Krebs-Thomson K,
    3. Braff DL, and
    4. Swerdlow NR
    (2001) Pharmacological studies of prepulse inhibition models of sensorimotor gating deficits in schizophrenia: a decade in review. Psychopharmacology (Berl) 156:117–154.
    OpenUrlCrossRefPubMed
  45. ↵
    1. Gilmour G,
    2. Dix S,
    3. Fellini L,
    4. Gastambide F,
    5. Plath N,
    6. Steckler T,
    7. Talpos J, and
    8. Tricklebank M
    (2012) NMDA receptors, cognition and schizophrenia--testing the validity of the NMDA receptor hypofunction hypothesis. Neuropharmacology 62:1401–1412.
    OpenUrlCrossRefPubMed
  46. ↵
    1. Gordon JA
    (2010) Testing the glutamate hypothesis of schizophrenia. Nat Neurosci 13:2–4.
    OpenUrlCrossRefPubMed
  47. ↵
    1. Gould RW,
    2. Dencker D,
    3. Grannan M,
    4. Bubser M,
    5. Zhan X,
    6. Wess J,
    7. Xiang Z,
    8. Locuson C,
    9. Lindsley CW,
    10. Conn PJ,
    11. et al.
    (2015) Role for the M1 muscarinic acetylcholine receptor in top-down cognitive processing using a touchscreen visual discrimination task in mice. ACS Chem Neurosci 6:1683–1695.
    OpenUrlCrossRef
  48. ↵
    1. Gronier B,
    2. Perry KW, and
    3. Rasmussen K
    (2000) Activation of the mesocorticolimbic dopaminergic system by stimulation of muscarinic cholinergic receptors in the ventral tegmental area. Psychopharmacology (Berl) 147:347–355.
    OpenUrlCrossRefPubMed
  49. ↵
    1. Grube BS,
    2. Bilder RM, and
    3. Goldman RS
    (1998) Meta-analysis of symptom factors in schizophrenia. Schizophr Res 31:113–120.
    OpenUrlCrossRefPubMed
  50. ↵
    1. Han M,
    2. Newell K,
    3. Zavitsanou K,
    4. Deng C, and
    5. Huang XF
    (2008) Effects of antipsychotic medication on muscarinic M1 receptor mRNA expression in the rat brain. J Neurosci Res 86:457–464.
    OpenUrlCrossRefPubMed
  51. ↵
    1. Harrison PJ
    (2004) The hippocampus in schizophrenia: a review of the neuropathological evidence and its pathophysiological implications. Psychopharmacology (Berl) 174:151–162.
    OpenUrlCrossRefPubMed
  52. ↵
    1. Harrison PJ and
    2. Weinberger DR
    (2005) Schizophrenia genes, gene expression, and neuropathology: on the matter of their convergence. Mol Psychiatry 10:40–68; image 45.
    OpenUrlCrossRefPubMed
  53. ↵
    1. Harvey PD,
    2. Howanitz E,
    3. Parrella M,
    4. White L,
    5. Davidson M,
    6. Mohs RC,
    7. Hoblyn J, and
    8. Davis KL
    (1998) Symptoms, cognitive functioning, and adaptive skills in geriatric patients with lifelong schizophrenia: a comparison across treatment sites. Am J Psychiatry 155:1080–1086.
    OpenUrlCrossRefPubMed
  54. ↵
    1. Heckers S,
    2. Rauch SL,
    3. Goff D,
    4. Savage CR,
    5. Schacter DL,
    6. Fischman AJ, and
    7. Alpert NM
    (1998) Impaired recruitment of the hippocampus during conscious recollection in schizophrenia. Nat Neurosci 1:318–323.
    OpenUrlCrossRefPubMed
  55. ↵
    1. Heinrichs RW and
    2. Zakzanis KK
    (1998) Neurocognitive deficit in schizophrenia: a quantitative review of the evidence. Neuropsychology 12:426–445.
    OpenUrlCrossRefPubMed
  56. ↵
    1. Ishii D,
    2. Matsuzawa D,
    3. Kanahara N,
    4. Matsuda S,
    5. Sutoh C,
    6. Ohtsuka H,
    7. Nakazawa K,
    8. Kohno M,
    9. Hashimoto K,
    10. Iyo M,
    11. et al.
    (2010) Effects of aripiprazole on MK-801-induced prepulse inhibition deficits and mitogen-activated protein kinase signal transduction pathway. Neurosci Lett 471:53–57.
    OpenUrlCrossRefPubMed
  57. ↵
    1. Javitt DC
    (2007) Glutamate and schizophrenia: phencyclidine, N-methyl-D-aspartate receptors, and dopamine-glutamate interactions. Int Rev Neurobiol 78:69–108.
    OpenUrlCrossRefPubMed
  58. ↵
    1. Javitt DC and
    2. Zukin SR
    (1991) Recent advances in the phencyclidine model of schizophrenia. Am J Psychiatry 148:1301–1308.
    OpenUrlCrossRefPubMed
  59. ↵
    1. Jones CK,
    2. Brady AE,
    3. Davis AA,
    4. Xiang Z,
    5. Bubser M,
    6. Tantawy MN,
    7. Kane AS,
    8. Bridges TM,
    9. Kennedy JP,
    10. Bradley SR,
    11. et al.
    (2008) Novel selective allosteric activator of the M1 muscarinic acetylcholine receptor regulates amyloid processing and produces antipsychotic-like activity in rats. J Neurosci 28:10422–10433.
    OpenUrlAbstract/FREE Full Text
  60. ↵
    1. Kandel ER,
    2. Schwartz JH, and
    3. Jessell TM
    1. Kandel ER
    (2000) Disorders of thought and volition: schizophrenia, in Principles of Neural Science (Kandel ER, Schwartz JH, and Jessell TM eds) p 1414, McGraw-Hill, New York.
  61. ↵
    1. Kapur S,
    2. VanderSpek SC,
    3. Brownlee BA, and
    4. Nobrega JN
    (2003) Antipsychotic dosing in preclinical models is often unrepresentative of the clinical condition: a suggested solution based on in vivo occupancy. J Pharmacol Exp Ther 305:625–631.
    OpenUrlAbstract/FREE Full Text
  62. ↵
    1. Katsnelson A
    (2014) Drug development: The modelling challenge. Nature 508:S8–S9.
    OpenUrlCrossRef
  63. ↵
    1. Keefe RS,
    2. Bilder RM,
    3. Davis SM,
    4. Harvey PD,
    5. Palmer BW,
    6. Gold JM,
    7. Meltzer HY,
    8. Green MF,
    9. Capuano G,
    10. Stroup TS,
    11. et al.,
    12. CATIE Investigators, and
    13. Neurocognitive Working Group
    (2007) Neurocognitive effects of antipsychotic medications in patients with chronic schizophrenia in the CATIE Trial. Arch Gen Psychiatry 64:633–647.
    OpenUrlCrossRefPubMed
  64. ↵
    1. Keith VA,
    2. Mansbach RS, and
    3. Geyer MA
    (1991) Failure of haloperidol to block the effects of phencyclidine and dizocilpine on prepulse inhibition of startle. Biol Psychiatry 30:557–566.
    OpenUrlCrossRefPubMed
  65. ↵
    1. Kruse AC,
    2. Kobilka BK,
    3. Gautam D,
    4. Sexton PM,
    5. Christopoulos A, and
    6. Wess J
    (2014) Muscarinic acetylcholine receptors: novel opportunities for drug development. Nat Rev Drug Discov 13:549–560.
    OpenUrlCrossRefPubMed
  66. ↵
    1. Langmead CJ,
    2. Watson J, and
    3. Reavill C
    (2008) Muscarinic acetylcholine receptors as CNS drug targets. Pharmacol Ther 117:232–243.
    OpenUrlCrossRefPubMed
  67. ↵
    1. Lee I and
    2. Kesner RP
    (2003) Time-dependent relationship between the dorsal hippocampus and the prefrontal cortex in spatial memory. J Neurosci 23:1517–1523.
    OpenUrlAbstract/FREE Full Text
  68. ↵
    1. Lester DB,
    2. Rogers TD, and
    3. Blaha CD
    (2010) Acetylcholine-dopamine interactions in the pathophysiology and treatment of CNS disorders. CNS Neurosci Ther 16:137–162.
    OpenUrlCrossRefPubMed
  69. ↵
    1. Lieberman JA,
    2. Javitch JA, and
    3. Moore H
    (2008) Cholinergic agonists as novel treatments for schizophrenia: the promise of rational drug development for psychiatry. Am J Psychiatry 165:931–936.
    OpenUrlCrossRefPubMed
  70. ↵
    1. Lieberman JA,
    2. Stroup TS,
    3. McEvoy JP,
    4. Swartz MS,
    5. Rosenheck RA,
    6. Perkins DO,
    7. Keefe RS,
    8. Davis SM,
    9. Davis CE,
    10. Lebowitz BD,
    11. et al., and
    12. Clinical Antipsychotic Trials of Intervention Effectiveness (CATIE) Investigators
    (2005) Effectiveness of antipsychotic drugs in patients with chronic schizophrenia. N Engl J Med 353:1209–1223.
    OpenUrlCrossRefPubMed
  71. ↵
    1. Lipska BK and
    2. Weinberger DR
    (2002) A neurodevelopmental model of schizophrenia: neonatal disconnection of the hippocampus. Neurotox Res 4:469–475.
    OpenUrlCrossRefPubMed
  72. ↵
    1. Long LE,
    2. Malone DT, and
    3. Taylor DA
    (2006) Cannabidiol reverses MK-801-induced disruption of prepulse inhibition in mice. Neuropsychopharmacology 31:795–803.
    OpenUrlCrossRefPubMed
  73. ↵
    1. Lublin H,
    2. Eberhard J, and
    3. Levander S
    (2005) Current therapy issues and unmet clinical needs in the treatment of schizophrenia: a review of the new generation antipsychotics. Int Clin Psychopharmacol 20:183–198.
    OpenUrlCrossRefPubMed
  74. ↵
    1. Ma L,
    2. Seager MA,
    3. Wittmann M,
    4. Jacobson M,
    5. Bickel D,
    6. Burno M,
    7. Jones K,
    8. Graufelds VK,
    9. Xu G,
    10. Pearson M,
    11. et al.
    (2009) Selective activation of the M1 muscarinic acetylcholine receptor achieved by allosteric potentiation. Proc Natl Acad Sci USA 106:15950–15955.
    OpenUrlAbstract/FREE Full Text
  75. ↵
    1. Mansbach RS,
    2. Geyer MA, and
    3. Braff DL
    (1988) Dopaminergic stimulation disrupts sensorimotor gating in the rat. Psychopharmacology (Berl) 94:507–514.
    OpenUrlCrossRefPubMed
  76. ↵
    1. Matsuo N,
    2. Takao K,
    3. Nakanishi K,
    4. Yamasaki N,
    5. Tanda K, and
    6. Miyakawa T
    (2010) Behavioral profiles of three C57BL/6 substrains. Front Behav Neurosci 4:29.
    OpenUrlPubMed
  77. ↵
    1. Mauck B,
    2. Lucot JB,
    3. Paton S, and
    4. Grubbs RD
    (2010) Cholinesterase inhibitors and stress: effects on brain muscarinic receptor density in mice. Neurotoxicology 31:461–467.
    OpenUrlCrossRef
  78. ↵
    1. Miyamoto S,
    2. Duncan GE,
    3. Marx CE, and
    4. Lieberman JA
    (2005) Treatments for schizophrenia: a critical review of pharmacology and mechanisms of action of antipsychotic drugs. Mol Psychiatry 10:79–104.
    OpenUrlCrossRefPubMed
  79. ↵
    1. Miyamoto S,
    2. Miyake N,
    3. Jarskog LF,
    4. Fleischhacker WW, and
    5. Lieberman JA
    (2012) Pharmacological treatment of schizophrenia: a critical review of the pharmacology and clinical effects of current and future therapeutic agents. Mol Psychiatry 17:1206–1227.
    OpenUrlCrossRefPubMed
  80. ↵
    1. Moghaddam B and
    2. Javitt D
    (2012) From revolution to evolution: the glutamate hypothesis of schizophrenia and its implication for treatment. Neuropsychopharmacology 37:4–15.
    OpenUrlCrossRefPubMed
  81. ↵
    1. Moore H,
    2. Geyer MA,
    3. Carter CS, and
    4. Barch DM
    (2013) Harnessing cognitive neuroscience to develop new treatments for improving cognition in schizophrenia: CNTRICS selected cognitive paradigms for animal models. Neurosci Biobehav Rev 37 (Pt B):2087–2091.
    OpenUrlCrossRefPubMed
  82. ↵
    1. Murray JB
    (2002) Phencyclidine (PCP): a dangerous drug, but useful in schizophrenia research. J Psychol 136:319–327.
    OpenUrlCrossRefPubMed
  83. ↵
    1. Mutlu O,
    2. Ulak G,
    3. Celikyurt IK,
    4. Akar FY, and
    5. Erden F
    (2011) Effects of olanzapine, sertindole and clozapine on learning and memory in the Morris water maze test in naive and MK-801-treated mice. Pharmacol Biochem Behav 98:398–404.
    OpenUrlCrossRefPubMed
  84. ↵
    1. Nasrallah HA
    (2008) Atypical antipsychotic-induced metabolic side effects: insights from receptor-binding profiles. Mol Psychiatry 13:27–35.
    OpenUrlCrossRefPubMed
  85. ↵
    1. Natesan S,
    2. Reckless GE,
    3. Barlow KB,
    4. Nobrega JN, and
    5. Kapur S
    (2011) Partial agonists in schizophrenia--why some work and others do not: insights from preclinical animal models. Int J Neuropsychopharmacol 14:1165–1178.
    OpenUrlAbstract/FREE Full Text
  86. ↵
    1. Newell KA,
    2. Zavitsanou K, and
    3. Huang XF
    (2007) Opposing short- and long-term effects on muscarinic M1/4 receptor binding following chronic phencyclidine treatment. J Neurosci Res 85:1358–1363.
    OpenUrlCrossRefPubMed
  87. ↵
    1. Nicolazzo JA,
    2. Steuten JA,
    3. Charman SA,
    4. Taylor N,
    5. Davies PJ, and
    6. Petrou S
    (2010) Brain uptake of diazepam and phenytoin in a genetic animal model of absence epilepsy. Clin Exp Pharmacol Physiol 37:647–649.
    OpenUrlCrossRefPubMed
  88. ↵
    1. Nilsson M,
    2. Hansson S,
    3. Carlsson A, and
    4. Carlsson ML
    (2007) Differential effects of the N-methyl-d-aspartate receptor antagonist MK-801 on different stages of object recognition memory in mice. Neuroscience 149:123–130.
    OpenUrlCrossRefPubMed
  89. ↵
    1. Owen MJ,
    2. Sawa A, and
    3. Mortensen PB
    (2016) Schizophrenia. Lancet 388:86–97.
    OpenUrlCrossRef
  90. ↵
    1. Pantelis C,
    2. Barber FZ,
    3. Barnes TR,
    4. Nelson HE,
    5. Owen AM, and
    6. Robbins TW
    (1999) Comparison of set-shifting ability in patients with chronic schizophrenia and frontal lobe damage. Schizophr Res 37:251–270.
    OpenUrlCrossRefPubMed
  91. ↵
    1. Pantelis C and
    2. Barnes TR
    (1996) Drug strategies and treatment-resistant schizophrenia. Aust N Z J Psychiatry 30:20–37.
    OpenUrlAbstract/FREE Full Text
  92. ↵
    1. Pantelis C,
    2. Barnes TR,
    3. Nelson HE,
    4. Tanner S,
    5. Weatherley L,
    6. Owen AM, and
    7. Robbins TW
    (1997) Frontal-striatal cognitive deficits in patients with chronic schizophrenia. Brain 120:1823–1843.
    OpenUrlAbstract/FREE Full Text
  93. ↵
    1. Pantelis C and
    2. Lambert TJ
    (2003) Managing patients with “treatment-resistant” schizophrenia. Med J Aust 178 (Suppl):S62–S66.
    OpenUrlPubMed
  94. ↵
    1. Pantelis C,
    2. Stuart GW,
    3. Nelson HE,
    4. Robbins TW, and
    5. Barnes TR
    (2001) Spatial working memory deficits in schizophrenia: relationship with tardive dyskinesia and negative symptoms. Am J Psychiatry 158:1276–1285.
    OpenUrlCrossRefPubMed
  95. ↵
    1. Park SJ,
    2. Lee Y,
    3. Oh HK,
    4. Lee HE,
    5. Lee Y,
    6. Ko SY,
    7. Kim B,
    8. Cheong JH,
    9. Shin CY, and
    10. Ryu JH
    (2014) Oleanolic acid attenuates MK-801-induced schizophrenia-like behaviors in mice. Neuropharmacology 86:49–56.
    OpenUrlCrossRefPubMed
  96. ↵
    1. Peng S,
    2. Zhang Y,
    3. Li GJ,
    4. Zhang DX,
    5. Sun DP, and
    6. Fang Q
    (2012) The effect of sevoflurane on the expression of M1 acetylcholine receptor in the hippocampus and cognitive function of aged rats. Mol Cell Biochem 361:229–233.
    OpenUrlCrossRefPubMed
  97. ↵
    1. Pennartz CM,
    2. Ito R,
    3. Verschure PF,
    4. Battaglia FP, and
    5. Robbins TW
    (2011) The hippocampal-striatal axis in learning, prediction and goal-directed behavior. Trends Neurosci 34:548–559.
    OpenUrlCrossRefPubMed
  98. ↵
    1. Porter AC,
    2. Bymaster FP,
    3. DeLapp NW,
    4. Yamada M,
    5. Wess J,
    6. Hamilton SE,
    7. Nathanson NM, and
    8. Felder CC
    (2002) M1 muscarinic receptor signaling in mouse hippocampus and cortex. Brain Res 944:82–89.
    OpenUrlCrossRefPubMed
  99. ↵
    1. Roth BL,
    2. Sheffler DJ, and
    3. Kroeze WK
    (2004) Magic shotguns versus magic bullets: selectively non-selective drugs for mood disorders and schizophrenia. Nat Rev Drug Discov 3:353–359.
    OpenUrlCrossRefPubMed
  100. ↵
    1. Rujescu D,
    2. Bender A,
    3. Keck M,
    4. Hartmann AM,
    5. Ohl F,
    6. Raeder H,
    7. Giegling I,
    8. Genius J,
    9. McCarley RW,
    10. Möller HJ,
    11. et al.
    (2006) A pharmacological model for psychosis based on N-methyl-D-aspartate receptor hypofunction: molecular, cellular, functional and behavioral abnormalities. Biol Psychiatry 59:721–729.
    OpenUrlCrossRefPubMed
  101. ↵
    1. Sakurai H,
    2. Bies RR,
    3. Stroup ST,
    4. Keefe RS,
    5. Rajji TK,
    6. Suzuki T,
    7. Mamo DC,
    8. Pollock BG,
    9. Watanabe K,
    10. Mimura M,
    11. et al.
    (2013) Dopamine D2 receptor occupancy and cognition in schizophrenia: analysis of the CATIE data. Schizophr Bull 39:564–574.
    OpenUrlAbstract/FREE Full Text
  102. ↵
    1. Saykin AJ,
    2. Shtasel DL,
    3. Gur RE,
    4. Kester DB,
    5. Mozley LH,
    6. Stafiniak P, and
    7. Gur RC
    (1994) Neuropsychological deficits in neuroleptic naive patients with first-episode schizophrenia. Arch Gen Psychiatry 51:124–131.
    OpenUrlCrossRefPubMed
  103. ↵
    1. Shekhar A,
    2. Potter WZ,
    3. Lightfoot J,
    4. Lienemann J,
    5. Dubé S,
    6. Mallinckrodt C,
    7. Bymaster FP,
    8. McKinzie DL, and
    9. Felder CC
    (2008) Selective muscarinic receptor agonist xanomeline as a novel treatment approach for schizophrenia. Am J Psychiatry 165:1033–1039.
    OpenUrlCrossRefPubMed
  104. ↵
    1. Shipton OA,
    2. El-Gaby M,
    3. Apergis-Schoute J,
    4. Deisseroth K,
    5. Bannerman DM,
    6. Paulsen O, and
    7. Kohl MM
    (2014) Left-right dissociation of hippocampal memory processes in mice. Proc Natl Acad Sci USA 111:15238–15243.
    OpenUrlAbstract/FREE Full Text
  105. ↵
    1. Shirey JK,
    2. Brady AE,
    3. Jones PJ,
    4. Davis AA,
    5. Bridges TM,
    6. Kennedy JP,
    7. Jadhav SB,
    8. Menon UN,
    9. Xiang Z,
    10. Watson ML,
    11. et al.
    (2009) A selective allosteric potentiator of the M1 muscarinic acetylcholine receptor increases activity of medial prefrontal cortical neurons and restores impairments in reversal learning. J Neurosci 29:14271–14286.
    OpenUrlAbstract/FREE Full Text
  106. ↵
    1. Speller JC,
    2. Barnes TR,
    3. Curson DA,
    4. Pantelis C, and
    5. Alberts JL
    (1997) One-year, low-dose neuroleptic study of in-patients with chronic schizophrenia characterised by persistent negative symptoms. Amisulpride v. haloperidol. Br J Psychiatry 171:564–568.
    OpenUrlAbstract/FREE Full Text
  107. ↵
    1. Stone JM,
    2. Morrison PD, and
    3. Pilowsky LS
    (2007) Glutamate and dopamine dysregulation in schizophrenia--a synthesis and selective review. J Psychopharmacol 21:440–452.
    OpenUrlAbstract/FREE Full Text
  108. ↵
    1. Suratman S,
    2. Leach K,
    3. Sexton P,
    4. Felder C,
    5. Loiacono R, and
    6. Christopoulos A
    (2011) Impact of species variability and ‘probe-dependence’ on the detection and in vivo validation of allosteric modulation at the M4 muscarinic acetylcholine receptor. Br J Pharmacol 162:1659–1670.
    OpenUrlCrossRefPubMed
  109. ↵
    1. Sweatt JD
    (2004) Hippocampal function in cognition. Psychopharmacology (Berl) 174:99–110.
    OpenUrlCrossRefPubMed
  110. ↵
    1. Swerdlow NR,
    2. Braff DL, and
    3. Geyer MA
    (1990) GABAergic projection from nucleus accumbens to ventral pallidum mediates dopamine-induced sensorimotor gating deficits of acoustic startle in rats. Brain Res 532:146–150.
    OpenUrlCrossRefPubMed
  111. ↵
    1. Swerdlow NR,
    2. Braff DL,
    3. Taaid N, and
    4. Geyer MA
    (1994) Assessing the validity of an animal model of deficient sensorimotor gating in schizophrenic patients. Arch Gen Psychiatry 51:139–154.
    OpenUrlCrossRefPubMed
  112. ↵
    1. Swerdlow NR and
    2. Geyer MA
    (1998) Using an animal model of deficient sensorimotor gating to study the pathophysiology and new treatments of schizophrenia. Schizophr Bull 24:285–301.
    OpenUrlAbstract/FREE Full Text
  113. ↵
    1. Swerdlow NR,
    2. Geyer MA, and
    3. Braff DL
    (2001) Neural circuit regulation of prepulse inhibition of startle in the rat: current knowledge and future challenges. Psychopharmacology (Berl) 156:194–215.
    OpenUrlCrossRefPubMed
  114. ↵
    1. Talts U,
    2. Talts JF, and
    3. Eriksson P
    (1998) Differential expression of muscarinic subtype mRNAs after exposure to neurotoxic pesticides. Neurobiol Aging 19:553–559.
    OpenUrlCrossRefPubMed
  115. ↵
    1. Tanskanen P,
    2. Veijola JM,
    3. Piippo UK,
    4. Haapea M,
    5. Miettunen JA,
    6. Pyhtinen J,
    7. Bullmore ET,
    8. Jones PB, and
    9. Isohanni MK
    (2005) Hippocampus and amygdala volumes in schizophrenia and other psychoses in the Northern Finland 1966 birth cohort. Schizophr Res 75:283–294.
    OpenUrlCrossRefPubMed
  116. ↵
    1. Thal DM,
    2. Sun B,
    3. Feng D,
    4. Nawaratne V,
    5. Leach K,
    6. Felder CC,
    7. Bures MG,
    8. Evans DA,
    9. Weis WI,
    10. Bachhawat P,
    11. et al.
    (2016) Crystal structures of the M1 and M4 muscarinic acetylcholine receptors. Nature 531:335–340.
    OpenUrlCrossRefPubMed
  117. ↵
    1. Thomsen M,
    2. Wess J,
    3. Fulton BS,
    4. Fink-Jensen A, and
    5. Caine SB
    (2010) Modulation of prepulse inhibition through both M(1) and M (4) muscarinic receptors in mice. Psychopharmacology (Berl) 208:401–416.
    OpenUrlCrossRef
  118. ↵
    1. Turnock M and
    2. Becker S
    (2008) A neural network model of hippocampal-striatal-prefrontal interactions in contextual conditioning. Brain Res 1202:87–98.
    OpenUrlCrossRefPubMed
  119. ↵
    1. Vakalopoulos C
    (2006) Neuropharmacology of cognition and memory: a unifying theory of neuromodulator imbalance in psychiatry and amnesia. Med Hypotheses 66:394–431.
    OpenUrlCrossRefPubMed
  120. ↵
    1. van der Staay FJ,
    2. Rutten K,
    3. Erb C, and
    4. Blokland A
    (2011) Effects of the cognition impairer MK-801 on learning and memory in mice and rats. Behav Brain Res 220:215–229.
    OpenUrlCrossRefPubMed
  121. ↵
    1. van Erp TG,
    2. Hibar DP,
    3. Rasmussen JM,
    4. Glahn DC,
    5. Pearlson GD,
    6. Andreassen OA,
    7. Agartz I,
    8. Westlye LT,
    9. Haukvik UK,
    10. Dale AM,
    11. et al.
    (2016) Subcortical brain volume abnormalities in 2028 individuals with schizophrenia and 2540 healthy controls via the ENIGMA consortium. Mol Psychiatry 21:547–553.
    OpenUrlCrossRefPubMed
  122. ↵
    1. van Erp TG,
    2. Saleh PA,
    3. Huttunen M,
    4. Lönnqvist J,
    5. Kaprio J,
    6. Salonen O,
    7. Valanne L,
    8. Poutanen VP,
    9. Standertskjöld-Nordenstam CG, and
    10. Cannon TD
    (2004) Hippocampal volumes in schizophrenic twins. Arch Gen Psychiatry 61:346–353.
    OpenUrlCrossRefPubMed
  123. ↵
    1. Velakoulis D,
    2. Wood SJ,
    3. Wong MT,
    4. McGorry PD,
    5. Yung A,
    6. Phillips L,
    7. Smith D,
    8. Brewer W,
    9. Proffitt T,
    10. Desmond P,
    11. et al.
    (2006) Hippocampal and amygdala volumes according to psychosis stage and diagnosis: a magnetic resonance imaging study of chronic schizophrenia, first-episode psychosis, and ultra-high-risk individuals. Arch Gen Psychiatry 63:139–149.
    OpenUrlCrossRefPubMed
  124. ↵
    1. Weinberger DR
    (1999) Cell biology of the hippocampal formation in schizophrenia. Biol Psychiatry 45:395–402.
    OpenUrlCrossRefPubMed
  125. ↵
    1. Weinberger DR and
    2. Gallhofer B
    (1997) Cognitive function in schizophrenia. Int Clin Psychopharmacol 12 (Suppl 4):S29–S36.
    OpenUrlCrossRef
  126. ↵
    1. Wood MD,
    2. Murkitt KL,
    3. Ho M,
    4. Watson JM,
    5. Brown F,
    6. Hunter AJ, and
    7. Middlemiss DN
    (1999) Functional comparison of muscarinic partial agonists at muscarinic receptor subtypes hM1, hM2, hM3, hM4 and hM5 using microphysiometry. Br J Pharmacol 126:1620–1624.
    OpenUrlCrossRefPubMed
  127. ↵
    1. Wood SJ,
    2. Pantelis C,
    3. Proffitt T,
    4. Phillips LJ,
    5. Stuart GW,
    6. Buchanan JA,
    7. Mahony K,
    8. Brewer W,
    9. Smith DJ, and
    10. McGorry PD
    (2003) Spatial working memory ability is a marker of risk-for-psychosis. Psychol Med 33:1239–1247.
    OpenUrlCrossRefPubMed
  128. ↵
    1. Wood SJ,
    2. Proffitt T,
    3. Mahony K,
    4. Smith DJ,
    5. Buchanan JA,
    6. Brewer W,
    7. Stuart GW,
    8. Velakoulis D,
    9. McGorry PD, and
    10. Pantelis C
    (2002) Visuospatial memory and learning in first-episode schizophreniform psychosis and established schizophrenia: a functional correlate of hippocampal pathology? Psychol Med 32:429–438.
    OpenUrlCrossRefPubMed
  129. ↵
    1. Yee BK,
    2. Chang T,
    3. Pietropaolo S, and
    4. Feldon J
    (2005) The expression of prepulse inhibition of the acoustic startle reflex as a function of three pulse stimulus intensities, three prepulse stimulus intensities, and three levels of startle responsiveness in C57BL6/J mice. Behav Brain Res 163:265–276.
    OpenUrlCrossRefPubMed
  130. ↵
    1. Young JW,
    2. Zhou X, and
    3. Geyer MA
    (2010) Animal models of schizophrenia. Curr Top Behav Neurosci 4:391–433.
    OpenUrlCrossRefPubMed
  131. ↵
    1. Zhang WN,
    2. Bast T, and
    3. Feldon J
    (2000) Microinfusion of the non-competitive N-methyl-D-aspartate receptor antagonist MK-801 (dizocilpine) into the dorsal hippocampus of Wistar rats does not affect latent inhibition and prepulse inhibition, but increases startle reaction and locomotor activity. Neuroscience 101:589–599.
    OpenUrlCrossRefPubMed
PreviousNext
Back to top

In this issue

Journal of Pharmacology and Experimental Therapeutics: 359 (2)
Journal of Pharmacology and Experimental Therapeutics
Vol. 359, Issue 2
1 Nov 2016
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Index by author
  • Editorial Board (PDF)
  • Front Matter (PDF)
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Journal of Pharmacology and Experimental Therapeutics article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Positive Allosteric Modulation of the Muscarinic M1 Receptor Improves Efficacy of Antipsychotics in Mouse Glutamatergic Deficit Models of Behavior
(Your Name) has forwarded a page to you from Journal of Pharmacology and Experimental Therapeutics
(Your Name) thought you would be interested in this article in Journal of Pharmacology and Experimental Therapeutics.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Research ArticleDrug Discovery and Translational Medicine

M1 Positive Allosteric Modulation Improves Antipsychotic Efficacy

Kwok H. C. Choy, David M. Shackleford, Daniel T. Malone, Shailesh N. Mistry, Rahul T. Patil, Peter J. Scammells, Christopher J. Langmead, Christos Pantelis, Patrick M. Sexton, Johnathan R. Lane and Arthur Christopoulos
Journal of Pharmacology and Experimental Therapeutics November 1, 2016, 359 (2) 354-365; DOI: https://doi.org/10.1124/jpet.116.235788

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Research ArticleDrug Discovery and Translational Medicine

M1 Positive Allosteric Modulation Improves Antipsychotic Efficacy

Kwok H. C. Choy, David M. Shackleford, Daniel T. Malone, Shailesh N. Mistry, Rahul T. Patil, Peter J. Scammells, Christopher J. Langmead, Christos Pantelis, Patrick M. Sexton, Johnathan R. Lane and Arthur Christopoulos
Journal of Pharmacology and Experimental Therapeutics November 1, 2016, 359 (2) 354-365; DOI: https://doi.org/10.1124/jpet.116.235788
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Introduction
    • Materials and Methods
    • Results
    • Discussion
    • Acknowledgments
    • Authorship Contributions
    • Footnotes
    • Abbreviations
    • References
  • Figures & Data
  • Info & Metrics
  • eLetters
  • PDF + SI
  • PDF

Related Articles

Cited By...

More in this TOC Section

  • C. Butyricum Alleviates bone loss after Bariatric Surgery
  • Running title: Antimuscarinic effect of mirabegron
  • IKCa Channels in Muscle Hypertrophy
Show more Drug Discovery and Translational Medicine

Similar Articles

  • Home
  • Alerts
Facebook   Twitter   LinkedIn   RSS

Navigate

  • Current Issue
  • Fast Forward by date
  • Fast Forward by section
  • Latest Articles
  • Archive
  • Search for Articles
  • Feedback
  • ASPET

More Information

  • About JPET
  • Editorial Board
  • Instructions to Authors
  • Submit a Manuscript
  • Customized Alerts
  • RSS Feeds
  • Subscriptions
  • Permissions
  • Terms & Conditions of Use

ASPET's Other Journals

  • Drug Metabolism and Disposition
  • Molecular Pharmacology
  • Pharmacological Reviews
  • Pharmacology Research & Perspectives
ISSN 1521-0103 (Online)

Copyright © 2021 by the American Society for Pharmacology and Experimental Therapeutics